1
|
Marchese AM, Fries L, Beyhaghi H, Vadivale M, Zhu M, Cloney-Clark S, Plested JS, Chung AW, Dunkle LM, Kalkeri R. Mechanisms and implications of IgG4 responses to SARS-CoV-2 and other repeatedly administered vaccines. J Infect 2024; 89:106317. [PMID: 39419185 DOI: 10.1016/j.jinf.2024.106317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Vaccine-induced immunoglobulin G (IgG) profiles can vary with respect to the predominant subclasses that characterize the response. Among IgG subclasses, IgG4 is reported to have anti-inflammatory properties, but can also exhibit reduced capacity for virus neutralization and activation of Fc-dependent effector functions. Here, we review evidence that IgG4 subclass responses can be disproportionately increased in response to some types of vaccines targeting an array of diseases, including pertussis, HIV, malaria, and COVID-19. The basis for enhanced IgG4 induction by vaccines is poorly understood but may be associated with platform- or dose regimen-specific differences in antigen exposure and/or cytokine stimulation. The clinical implications of vaccine-induced IgG4 responses remain uncertain, though collective evidence suggests that proportional increases in IgG4 might reduce vaccine antigen-specific immunity. Additional work is needed to determine underlying mechanisms and to elucidate what role IgG4 may play in modifications of vaccine-induced immunity to disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Amy W Chung
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3000, Australia
| | | | | |
Collapse
|
2
|
Han J, Sheng T, Zhang Y, Cheng H, Gao J, Yu J, Gu Z. Bioresponsive Immunotherapeutic Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2209778. [PMID: 36639983 DOI: 10.1002/adma.202209778] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/31/2022] [Indexed: 06/17/2023]
Abstract
The human immune system is an interaction network of biological processes, and its dysfunction is closely associated with a wide array of diseases, such as cancer, infectious diseases, tissue damage, and autoimmune diseases. Manipulation of the immune response network in a desired and controlled fashion has been regarded as a promising strategy for maximizing immunotherapeutic efficacy and minimizing side effects. Integration of "smart" bioresponsive materials with immunoactive agents including small molecules, biomacromolecules, and cells can achieve on-demand release of agents at targeted sites to reduce overdose-related toxicity and alleviate off-target effects. This review highlights the design principles of bioresponsive immunotherapeutic materials and discusses the critical roles of controlled release of immunoactive agents from bioresponsive materials in recruiting, housing, and manipulating immune cells for evoking desired immune responses. Challenges and future directions from the perspective of clinical translation are also discussed.
Collapse
Affiliation(s)
- Jinpeng Han
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Tao Sheng
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hao Cheng
- Department of Materials Science and Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Jianqing Gao
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
3
|
Sircy LM, Ramstead AG, Gibbs LC, Joshi H, Baessler A, Mena I, García-Sastre A, Emerson LL, Fairfax KC, Williams MA, Hale JS. Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection. PLoS Pathog 2024; 20:e1011639. [PMID: 39283916 PMCID: PMC11404825 DOI: 10.1371/journal.ppat.1011639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 08/05/2024] [Indexed: 09/22/2024] Open
Abstract
Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection or immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.
Collapse
Affiliation(s)
- Linda M. Sircy
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew G. Ramstead
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Lisa C. Gibbs
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hemant Joshi
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew Baessler
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lyska L. Emerson
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Keke C. Fairfax
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - J. Scott Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
4
|
Elizaldi SR, Hawes CE, Verma A, Shaan Lakshmanappa Y, Dinasarapu AR, Schlegel BT, Rajasundaram D, Li J, Durbin-Johnson BP, Ma ZM, Pal PB, Beckman D, Ott S, Raeman R, Lifson J, Morrison JH, Iyer SS. Chronic SIV-Induced neuroinflammation disrupts CCR7+ CD4+ T cell immunosurveillance in the rhesus macaque brain. J Clin Invest 2024; 134:e175332. [PMID: 38470479 PMCID: PMC11060742 DOI: 10.1172/jci175332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024] Open
Abstract
CD4+ T cells survey and maintain immune homeostasis in the brain, yet their differentiation states and functional capabilities remain unclear. Our approach, combining single-cell transcriptomic analysis, ATAC-Seq, spatial transcriptomics, and flow cytometry, revealed a distinct subset of CCR7+ CD4+ T cells resembling lymph node central memory (TCM) cells. We observed chromatin accessibility at the CCR7, CD28, and BCL-6 loci, defining molecular features of TCM. Brain CCR7+ CD4+ T cells exhibited recall proliferation and interleukin-2 production ex vivo, showcasing their functional competence. We identified the skull bone marrow as a local niche for these cells alongside CNS border tissues. Sequestering TCM cells in lymph nodes using FTY720 led to reduced CCR7+ CD4+ T cell frequencies in the cerebrospinal fluid, accompanied by increased monocyte levels and soluble markers indicating immune activation. In macaques chronically infected with SIVCL757 and experiencing viral rebound due to cessation of antiretroviral therapy, a decrease in brain CCR7+ CD4+ T cells was observed, along with increased microglial activation and initiation of neurodegenerative pathways. Our findings highlight a role for CCR7+ CD4+ T cells in CNS immune surveillance, and their decline during chronic SIV highlights their responsiveness to neuroinflammation.
Collapse
Affiliation(s)
| | - Chase E. Hawes
- Graduate Group in Immunology, UCD, Davis, California, USA
| | - Anil Verma
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, USA
| | | | - Ashok R. Dinasarapu
- Department of Neurology, School of Medicine, Emory University, Atlanta, Georgia, USA
| | - Brent T. Schlegel
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jie Li
- Bioinformatics Core, UCD, Davis, California, USA
| | | | - Zhong-Min Ma
- California National Primate Research Center, UCD, Davis, California, USA
| | - Pabitra B. Pal
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, USA
| | - Danielle Beckman
- California National Primate Research Center, UCD, Davis, California, USA
| | - Sean Ott
- California National Primate Research Center, UCD, Davis, California, USA
| | - Reben Raeman
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, USA
| | - Jeffrey Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, USA
| | - John H. Morrison
- California National Primate Research Center, UCD, Davis, California, USA
- Department of Neurology, School of Medicine, and
| | - Smita S. Iyer
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, USA
- California National Primate Research Center, UCD, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UCD, Davis, California, USA
| |
Collapse
|
5
|
Verma A, Hawes CE, Elizaldi SR, Smith JC, Rajasundaram D, Pedersen GK, Shen X, Williams LD, Tomaras GD, Kozlowski PA, Amara RR, Iyer SS. Tailoring T fh profiles enhances antibody persistence to a clade C HIV-1 vaccine in rhesus macaques. eLife 2024; 12:RP89395. [PMID: 38385642 PMCID: PMC10942585 DOI: 10.7554/elife.89395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024] Open
Abstract
CD4 T follicular helper cells (Tfh) are essential for establishing serological memory and have distinct helper attributes that impact both the quantity and quality of the antibody response. Insights into Tfh subsets that promote antibody persistence and functional capacity can critically inform vaccine design. Based on the Tfh profiles evoked by the live attenuated measles virus vaccine, renowned for its ability to establish durable humoral immunity, we investigated the potential of a Tfh1/17 recall response during the boost phase to enhance persistence of HIV-1 Envelope (Env) antibodies in rhesus macaques. Using a DNA-prime encoding gp160 antigen and Tfh polarizing cytokines (interferon protein-10 (IP-10) and interleukin-6 (IL-6)), followed by a gp140 protein boost formulated in a cationic liposome-based adjuvant (CAF01), we successfully generated germinal center (GC) Tfh1/17 cells. In contrast, a similar DNA-prime (including IP-10) followed by gp140 formulated with monophosphoryl lipid A (MPLA) +QS-21 adjuvant predominantly induced GC Tfh1 cells. While the generation of GC Tfh1/17 cells with CAF01 and GC Tfh1 cells with MPLA +QS-21 induced comparable peak Env antibodies, the latter group demonstrated significantly greater antibody concentrations at week 8 after final immunization which persisted up to 30 weeks (gp140 IgG ng/ml- MPLA; 5500; CAF01, 2155; p<0.05). Notably, interferon γ+Env-specific Tfh responses were consistently higher with gp140 in MPLA +QS-21 and positively correlated with Env antibody persistence. These findings suggest that vaccine platforms maximizing GC Tfh1 induction promote persistent Env antibodies, important for protective immunity against HIV.
Collapse
Affiliation(s)
- Anil Verma
- Department of Pathology, School of Medicine, University of PittsburghPittsburghUnited States
| | - Chase E Hawes
- Graduate Group in Immunology, University of California, DavisDavisUnited States
- California National Primate Research Center, University of California, DavisDavisUnited States
| | - Sonny R Elizaldi
- Graduate Group in Immunology, University of California, DavisDavisUnited States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, DavisDavisUnited States
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences CenterNew OrleansUnited States
| | - Dhivyaa Rajasundaram
- Bioinformatics Core, Department of Pediatrics, UPMC Children's Hospital of PittsburghPittsburghUnited States
| | | | - Xiaoying Shen
- Center for Human Systems ImmunologyDurhamUnited States
- Department of Surgery, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke University Medical CenterDurhamUnited States
| | - LaTonya D Williams
- Center for Human Systems ImmunologyDurhamUnited States
- Department of Surgery, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke University Medical CenterDurhamUnited States
| | - Georgia D Tomaras
- Center for Human Systems ImmunologyDurhamUnited States
- Department of Surgery, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke University Medical CenterDurhamUnited States
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
- Department of Integrative Immunobiology, Duke University Medical CenterDurhamUnited States
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences CenterNew OrleansUnited States
| | - Rama R Amara
- Department of Microbiology and Immunology, Emory UniversityAtlantaUnited States
- Yerkes National Primate Research Center, Emory UniversityAtlantaUnited States
| | - Smita S Iyer
- Department of Pathology, School of Medicine, University of PittsburghPittsburghUnited States
- California National Primate Research Center, University of California, DavisDavisUnited States
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, DavisDavisUnited States
| |
Collapse
|
6
|
Elizaldi SR, Verma A, Ma ZM, Ott S, Rajasundaram D, Hawes CE, Lakshmanappa YS, Cottrell ML, Kashuba ADM, Ambrose Z, Lifson JD, Morrison JH, Iyer SS. Deep analysis of CD4 T cells in the rhesus CNS during SIV infection. PLoS Pathog 2023; 19:e1011844. [PMID: 38060615 PMCID: PMC10729971 DOI: 10.1371/journal.ppat.1011844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/19/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023] Open
Abstract
Virologic suppression with antiretroviral therapy (ART) has significantly improved health outcomes for people living with HIV, yet challenges related to chronic inflammation in the central nervous system (CNS)-known as Neuro-HIV- persist. As primary targets for HIV-1 with the ability to survey and populate the CNS and interact with myeloid cells to co-ordinate neuroinflammation, CD4 T cells are pivotal in Neuro-HIV. Despite their importance, our understanding of CD4 T cell distribution in virus-targeted CNS tissues, their response to infection, and potential recovery following initiation of ART remain limited. To address these gaps, we studied ten SIVmac251-infected rhesus macaques using an ART regimen simulating suboptimal adherence. We evaluated four macaques during the acute phase pre-ART and six during the chronic phase. Our data revealed that HIV target CCR5+ CD4 T cells inhabit both the brain parenchyma and adjacent CNS tissues, encompassing choroid plexus stroma, dura mater, and the skull bone marrow. Aligning with the known susceptibility of CCR5+ CD4 T cells to viral infection and their presence within the CNS, high levels of viral RNA were detected in the brain parenchyma and its border tissues during acute SIV infection. Single-cell RNA sequencing of CD45+ cells from the brain revealed colocalization of viral transcripts within CD4 clusters and significant activation of antiviral molecules and specific effector programs within T cells, indicating CNS CD4 T cell engagement during infection. Acute infection led to marked imbalance in the CNS CD4/CD8 ratio which persisted into the chronic phase. These observations underscore the functional involvement of CD4 T cells within the CNS during SIV infection, enhancing our understanding of their role in establishing CNS viral presence. Our findings offer insights for potential T cell-focused interventions while underscoring the challenges in eradicating HIV from the CNS, particularly in the context of sub-optimal ART.
Collapse
Affiliation(s)
- Sonny R. Elizaldi
- Graduate Group in Immunology, UC Davis, California, United States of America
| | - Anil Verma
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
| | - Zhong-Min Ma
- California National Primate Research Center, UC Davis, California, United States of America
| | - Sean Ott
- California National Primate Research Center, UC Davis, California, United States of America
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
| | - Chase E. Hawes
- Graduate Group in Immunology, UC Davis, California, United States of America
| | | | - Mackenzie L. Cottrell
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Angela D. M. Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Zandrea Ambrose
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, Maryland, United States of America
| | - John H. Morrison
- California National Primate Research Center, UC Davis, California, United States of America
- Department of Neurology, School of Medicine, UC Davis, California, United States of America
| | - Smita S. Iyer
- Department of Pathology, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
- California National Primate Research Center, UC Davis, California, United States of America
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, California, United States of America
| |
Collapse
|
7
|
Sircy LM, Ramstead AG, Joshi H, Baessler A, Mena I, García-Sastre A, Williams MA, Scott Hale J. Generation of antigen-specific memory CD4 T cells by heterologous immunization enhances the magnitude of the germinal center response upon influenza infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.29.555253. [PMID: 37693425 PMCID: PMC10491174 DOI: 10.1101/2023.08.29.555253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Current influenza vaccine strategies have yet to overcome significant obstacles, including rapid antigenic drift of seasonal influenza viruses, in generating efficacious long-term humoral immunity. Due to the necessity of germinal center formation in generating long-lived high affinity antibodies, the germinal center has increasingly become a target for the development of novel or improvement of less-efficacious vaccines. However, there remains a major gap in current influenza research to effectively target T follicular helper cells during vaccination to alter the germinal center reaction. In this study, we used a heterologous infection or immunization priming strategy to seed an antigen-specific memory CD4+ T cell pool prior to influenza infection in mice to evaluate the effect of recalled memory T follicular helper cells in increased help to influenza-specific primary B cells and enhanced generation of neutralizing antibodies. We found that heterologous priming with intranasal infection with acute lymphocytic choriomeningitis virus (LCMV) or intramuscular immunization with adjuvanted recombinant LCMV glycoprotein induced increased antigen-specific effector CD4+ T and B cellular responses following infection with a recombinant influenza strain that expresses LCMV glycoprotein. Heterologously primed mice had increased expansion of secondary Th1 and Tfh cell subsets, including increased CD4+ TRM cells in the lung. However, the early enhancement of the germinal center cellular response following influenza infection did not impact influenza-specific antibody generation or B cell repertoires compared to primary influenza infection. Overall, our study suggests that while heterologous infection/immunization priming of CD4+ T cells is able to enhance the early germinal center reaction, further studies to understand how to target the germinal center and CD4+ T cells specifically to increase long-lived antiviral humoral immunity are needed.
Collapse
Affiliation(s)
- Linda M. Sircy
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew G. Ramstead
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Hemant Joshi
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Andrew Baessler
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Matthew A. Williams
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| | - J. Scott Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
8
|
Elizaldi SR, Verma A, Ma ZM, Ott S, Rajasundaram D, Cottrell ML, Kashuba ADM, Ambrose Z, Lifson JD, Morrison JH, Iyer SS. CD4 T cell Responses in the CNS during SIV infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554055. [PMID: 37662237 PMCID: PMC10473718 DOI: 10.1101/2023.08.24.554055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Virologic suppression with antiretroviral therapy (ART) has significantly improved health outcomes for people living with HIV, yet challenges related to chronic inflammation in the central nervous system (CNS) - known as Neuro-HIV- persist. As primary targets for HIV-1 with the ability to survey and populate the CNS and interact with myeloid cells to co-ordinate neuroinflammation, CD4 T cells are pivotal in Neuro-HIV. Despite their importance, our understanding of CD4 T cell distribution in virus-targeted CNS tissues, their response to infection, and potential recovery following initiation of ART remain limited. To address these gaps, we studied ten SIVmac251-infected rhesus macaques using an ART regimen simulating suboptimal adherence. We evaluated four macaques during the acute phase pre-ART and six during the chronic phase. Our data revealed that HIV target CCR5+ CD4 T cells inhabit both the brain parenchyma and adjacent CNS tissues, encompassing choroid plexus stroma, dura mater, and the skull bone marrow. Aligning with the known susceptibility of CCR5+ CD4 T cells to viral infection and their presence within the CNS, high levels of viral RNA were detected in the brain parenchyma and its border tissues during acute SIV infection. Single-cell RNA sequencing of CD45+ cells from the brain revealed colocalization of viral transcripts within CD4 clusters and significant activation of antiviral molecules and specific effector programs within T cells, indicating CNS CD4 T cell engagement during infection. Despite viral suppression with ART, acute infection led to significant depletion of CNS CD4 T cells, persisting into the chronic phase. These findings underscore the functional involvement of CD4 T cells within the CNS during SIV infection, enhancing our understanding of their role in establishing CNS viral presence. Our results offer insights for potential T cell-focused interventions while also underscoring the challenges in eradicating HIV from the CNS, even with effective ART.
Collapse
Affiliation(s)
| | - Anil Verma
- Department of Pathology, School of Medicine, University of Pittsburgh, PA, USA
| | - Zhong-Min Ma
- California National Primate Research Center, UC Davis, CA, USA
| | - Sean Ott
- California National Primate Research Center, UC Davis, CA, USA
| | | | | | - Angela D M Kashuba
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Zandrea Ambrose
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, PA, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD, USA
| | - John H Morrison
- California National Primate Research Center, UC Davis, CA, USA
- Department of Neurology, School of Medicine, UC Davis, CA,USA
| | - Smita S Iyer
- Department of Pathology, School of Medicine, University of Pittsburgh, PA, USA
- California National Primate Research Center, UC Davis, CA, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA,USA
| |
Collapse
|
9
|
Costanzo MC, Paquin-Proulx D, Schuetz A, Akapirat S, Shubin Z, Kim D, Wieczorek L, Polonis VR, Trinh HV, Rao M, Anenia H, Barrera MD, Boeckelman J, Nails B, Thapa P, Zemil M, Sacdalan C, Kroon E, Kaewboon B, Tipsuk S, Jongrakthaitae S, Gurunathan S, Sinangil F, Kim JH, Robb ML, Ake JA, O'Connell RJ, Pitisutthithum P, Nitayaphan S, Chariyalertsak S, Eller MA, Phanuphak N, Vasan S. ALVAC-HIV and AIDSVAX B/E vaccination induce improved immune responses compared with AIDSVAX B/E vaccination alone. JCI Insight 2023; 8:167664. [PMID: 37154156 DOI: 10.1172/jci.insight.167664] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/09/2023] [Indexed: 05/10/2023] Open
Abstract
The RV144 phase III vaccine trial demonstrated that ALVAC-HIV and AIDSVAX B/E administration over 6 months resulted in 31% efficacy in preventing HIV acquisition, while administration of AIDSVAX B/E alone in both VAX003 and VAX004 studies failed to show efficacy. In this study, we aimed to understand the impact of ALVAC-HIV on the development of cellular, humoral, and functional immune responses compared to the administration of AIDSVAX B/E alone. ALVAC-HIV in combination with 3 doses of AIDSVAX B/E significantly increased CD4+ HIV-specific T cell responses, polyfunctionality, and proliferation compared with 3 doses of AIDSVAX B/E alone. Additionally, Env-specific plasmablasts and A244-specific memory B cells were identified with a significantly higher magnitude in the group that received ALVAC-HIV. Subsequently, data revealed increased magnitude of plasma IgG binding to and avidity for HIV Env in participants who received ALVAC-HIV compared with 3 doses of AIDSVAX B/E alone. Lastly, levels of the Fc-mediated effector functions antibody-dependent cellular cytotoxicity, NK cell activation, and trogocytosis were significantly increased in participants who received ALVAC-HIV compared with those receiving AIDSVAX B/E alone. Taken together, these results suggest that ALVAC-HIV plays an essential role in developing cellular and humoral immune responses to protein-boosted regimens relative to protein alone.
Collapse
Affiliation(s)
- Margaret C Costanzo
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Dominic Paquin-Proulx
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Alexandra Schuetz
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
- Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand
| | - Siriwat Akapirat
- Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand
| | - Zhanna Shubin
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Dohoon Kim
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Lindsay Wieczorek
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Victoria R Polonis
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hung V Trinh
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Mangala Rao
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Hanna Anenia
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Michael D Barrera
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Jacob Boeckelman
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Barbara Nails
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Pallavi Thapa
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Michelle Zemil
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Carlo Sacdalan
- SEARCH, Institution of HIV Research and Innovation, Bangkok, Thailand
| | - Eugene Kroon
- SEARCH, Institution of HIV Research and Innovation, Bangkok, Thailand
| | - Boot Kaewboon
- Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand
| | - Somporn Tipsuk
- Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand
| | | | | | - Faruk Sinangil
- Global Solutions for Infectious Diseases, South San Francisco, California, USA
| | - Jerome H Kim
- International Vaccine Institute, Seoul, South Korea
| | - Merlin L Robb
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Julie A Ake
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Robert J O'Connell
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Armed Forces Research Institute for Medical Sciences, Bangkok, Thailand
| | | | | | | | - Michael A Eller
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Nittaya Phanuphak
- SEARCH, Institution of HIV Research and Innovation, Bangkok, Thailand
| | - Sandhya Vasan
- The US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Rahman MA, Becerra-Flores M, Patskovsky Y, Silva de Castro I, Bissa M, Basu S, Shen X, Williams LD, Sarkis S, N’guessan KF, LaBranche C, Tomaras GD, Aye PP, Veazey R, Paquin-Proulx D, Rao M, Franchini G, Cardozo T. Cholera toxin B scaffolded, focused SIV V2 epitope elicits antibodies that influence the risk of SIV mac251 acquisition in macaques. Front Immunol 2023; 14:1139402. [PMID: 37153584 PMCID: PMC10160393 DOI: 10.3389/fimmu.2023.1139402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction An efficacious HIV vaccine will need to elicit a complex package of innate, humoral, and cellular immune responses. This complex package of responses to vaccine candidates has been studied and yielded important results, yet it has been a recurring challenge to determine the magnitude and protective effect of specific in vivo immune responses in isolation. We therefore designed a single, viral-spike-apical, epitope-focused V2 loop immunogen to reveal individual vaccine-elicited immune factors that contribute to protection against HIV/SIV. Method We generated a novel vaccine by incorporating the V2 loop B-cell epitope in the cholera toxin B (CTB) scaffold and compared two new immunization regimens to a historically protective 'standard' vaccine regimen (SVR) consisting of 2xDNA prime boosted with 2xALVAC-SIV and 1xΔV1gp120. We immunized a cohort of macaques with 5xCTB-V2c vaccine+alum intramuscularly simultaneously with topical intrarectal vaccination of CTB-V2c vaccine without alum (5xCTB-V2/alum). In a second group, we tested a modified version of the SVR consisting of 2xDNA prime and boosted with 1xALVAC-SIV and 2xALVAC-SIV+CTB-V2/alum, (DA/CTB-V2c/alum). Results In the absence of any other anti-viral antibodies, V2c epitope was highly immunogenic when incorporated in the CTB scaffold and generated highly functional anti-V2c antibodies in the vaccinated animals. 5xCTB-V2c/alum vaccination mediated non-neutralizing ADCC activity and efferocytosis, but produced low avidity, trogocytosis, and no neutralization of tier 1 virus. Furthermore, DA/CTB-V2c/alum vaccination also generated lower total ADCC activity, avidity, and neutralization compared to the SVR. These data suggest that the ΔV1gp120 boost in the SVR yielded more favorable immune responses than its CTB-V2c counterpart. Vaccination with the SVR generates CCR5- α4β7+CD4+ Th1, Th2, and Th17 cells, which are less likely to be infected by SIV/HIV and likely contributed to the protection afforded in this regimen. The 5xCTB-V2c/alum regimen likewise elicited higher circulating CCR5- α4β7+ CD4+ T cells and mucosal α4β7+ CD4+ T cells compared to the DA/CTB-V2c/alum regimen, whereas the first cell type was associated with reduced risk of viral acquisition. Conclusion Taken together, these data suggest that individual viral spike B-cell epitopes can be highly immunogenic and functional as isolated immunogens, although they might not be sufficient on their own to provide full protection against HIV/SIV infection.
Collapse
Affiliation(s)
- Mohammad Arif Rahman
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH Bethesda, MD, United States
| | - Manuel Becerra-Flores
- NYU Langone Health, New York University School of Medicine, New York, NY, United States
| | - Yury Patskovsky
- NYU Langone Health, New York University School of Medicine, New York, NY, United States
| | - Isabela Silva de Castro
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH Bethesda, MD, United States
| | - Massimiliano Bissa
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH Bethesda, MD, United States
| | - Shraddha Basu
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - LaTonya D. Williams
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Sarkis Sarkis
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH Bethesda, MD, United States
| | - Kombo F. N’guessan
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Celia LaBranche
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Georgia D. Tomaras
- Department of Surgery, Duke University School of Medicine, Durham, NC, United States
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, United States
| | - Pyone Pyone Aye
- Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Ronald Veazey
- Division of Comparative Pathology, Department of Pathology and Laboratory Medicine, Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, United States
| | - Dominic Paquin-Proulx
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Mangala Rao
- United States Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Genoveffa Franchini
- Animal Models and Retroviral Vaccines Section, National Cancer Institute, NIH Bethesda, MD, United States
| | - Timothy Cardozo
- NYU Langone Health, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
11
|
Shrivastava S, Carmen JM, Lu Z, Basu S, Sankhala RS, Chen WH, Nguyen P, Chang WC, King J, Corbitt C, Mayer S, Bolton JS, Anderson A, Swafford I, Terriquez GD, Trinh HV, Kim J, Jobe O, Paquin-Proulx D, Matyas GR, Gromowski GD, Currier JR, Bergmann-Leitner E, Modjarrad K, Michael NL, Joyce MG, Malloy AMW, Rao M. SARS-CoV-2 spike-ferritin-nanoparticle adjuvanted with ALFQ induces long-lived plasma cells and cross-neutralizing antibodies. NPJ Vaccines 2023; 8:43. [PMID: 36934088 PMCID: PMC10024299 DOI: 10.1038/s41541-023-00638-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/28/2023] [Indexed: 03/20/2023] Open
Abstract
This study demonstrates the impact of adjuvant on the development of T follicular helper (Tfh) and B cells, and their influence on antibody responses in mice vaccinated with SARS-CoV-2-spike-ferritin-nanoparticle (SpFN) adjuvanted with either Army Liposome Formulation containing QS-21 (SpFN + ALFQ) or Alhydrogel® (SpFN + AH). SpFN + ALFQ increased the size and frequency of germinal center (GC) B cells in the vaccine-draining lymph nodes and increased the frequency of antigen-specific naive B cells. A single vaccination with SpFN + ALFQ resulted in a higher frequency of IL-21-producing-spike-specific Tfh and GC B cells in the draining lymph nodes and spleen, S-2P protein-specific IgM and IgG antibodies, and elicitation of robust cross-neutralizing antibodies against SARS-CoV-2 variants as early as day 7, which was enhanced by a second vaccination. This was associated with the generation of high titer, high avidity binding antibodies. The third vaccination with SpFN + ALFQ elicited high levels of neutralizing antibodies against the Omicron variant. No cross-neutralizing antibodies against Omicron were induced with SpFN + AH. These findings highlight the importance of ALFQ in orchestrating early induction of antigen-specific Tfh and GC B cell responses and long-lived plasma cells in the bone marrow. The early engagement of S-2P specific naive B cells and high titer IgM antibodies shape the development of long-term neutralization breadth.
Collapse
Affiliation(s)
- Shikha Shrivastava
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Joshua M Carmen
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Zhongyan Lu
- Department of Pediatrics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Shraddha Basu
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Rajeshwer S Sankhala
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Wei-Hung Chen
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Phuong Nguyen
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - William C Chang
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jocelyn King
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Courtney Corbitt
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sandra Mayer
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jessica S Bolton
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, USA
| | - Alexander Anderson
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Oak Ridge Institute of Science and Education, Oak Ridge, TN, 37831, USA
| | - Isabella Swafford
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Guillermo D Terriquez
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Hung V Trinh
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Jiae Kim
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Ousman Jobe
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Dominic Paquin-Proulx
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Gary R Matyas
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Gregory D Gromowski
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Jeffrey R Currier
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Elke Bergmann-Leitner
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, USA
| | - Kayvon Modjarrad
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Nelson L Michael
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - M Gordon Joyce
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Allison M W Malloy
- Department of Pediatrics, F. Edward Hebert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Mangala Rao
- Laboratory of Adjuvant and Antigen Research, U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
- Center for Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
12
|
Roy V, Jung W, Linde C, Coates E, Ledgerwood J, Costner P, Yamshchikov G, Streeck H, Juelg B, Lauffenburger DA, Alter G. Differences in HPV-specific antibody Fc-effector functions following Gardasil® and Cervarix® vaccination. NPJ Vaccines 2023; 8:39. [PMID: 36922512 PMCID: PMC10017795 DOI: 10.1038/s41541-023-00628-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/17/2023] [Indexed: 03/17/2023] Open
Abstract
Gardasil® (Merck) and Cervarix® (GlaxoSmithKline) both provide protection against infection with Human Papillomavirus 16 (HPV16) and Human Papillomavirus 18 (HPV18), that account for around 70% of cervical cancers. Both vaccines have been shown to induce high levels of neutralizing antibodies and are known to protect against progression beyond cervical intraepithelial neoplasia grade 2 (CIN2+), although Cervarix® has been linked to enhanced protection from progression. However, beyond the transmission-blocking activity of neutralizing antibodies against HPV, no clear correlate of protection has been defined that may explain persistent control and clearance elicited by HPV vaccines. Beyond blocking, antibodies contribute to antiviral activity via the recruitment of the cytotoxic and opsonophagocytic power of the immune system. Thus, here, we used systems serology to comprehensively profile Gardasil®- and Cervarix®- induced antibody subclass, isotype, Fc-receptor binding, and Fc-effector functions against the HPV16 and HPV18 major capsid protein (L1). Overall, both vaccines induced robust functional humoral immune responses against both HPV16 and HPV18. However, Cervarix® elicited higher IgG3 and antibody-dependent complement activating responses, and an overall more coordinated response between HPV16 and 18 compared to Gardasil®, potentially related to the distinct adjuvants delivered with the vaccines. Thus, these data point to robust Fc-effector functions induced by both Gardasil® and Cervarix®, albeit with enhanced coordination observed with Cervarix®, potentially underlying immunological correlates of post-infection control of HPV.
Collapse
Affiliation(s)
- Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.,Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Wonyeong Jung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Caitlyn Linde
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Emily Coates
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julie Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pamela Costner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Galina Yamshchikov
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, Bonn, Germany
| | - Boris Juelg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
13
|
Hawes CE, Elizaldi SR, Beckman D, Diniz GB, Shaan Lakshmanappa Y, Ott S, Durbin-Johnson BP, Dinasarapu AR, Gompers A, Morrison JH, Iyer SS. Neuroinflammatory transcriptional programs induced in rhesus pre-frontal cortex white matter during acute SHIV infection. J Neuroinflammation 2022; 19:250. [PMID: 36203187 PMCID: PMC9535930 DOI: 10.1186/s12974-022-02610-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background Immunosurveillance of the central nervous system (CNS) is vital to resolve infection and injury. However, immune activation within the CNS in the setting of chronic viral infections, such as HIV-1, is strongly linked to progressive neurodegeneration and cognitive decline. Establishment of HIV-1 in the CNS early following infection underscores the need to delineate features of acute CNS immune activation, as these early inflammatory events may mediate neurodegenerative processes. Here, we focused on elucidating molecular programs of neuroinflammation in brain regions based on vulnerability to neuroAIDS and/or neurocognitive decline. To this end, we assessed transcriptional profiles within the subcortical white matter of the pre-frontal cortex (PFCw), as well as synapse dense regions from hippocampus, superior temporal cortex, and caudate nucleus, in rhesus macaques following infection with Simian/Human Immunodeficiency Virus (SHIV.C.CH505). Methods We performed RNA extraction and sequenced RNA isolated from 3 mm brain punches. Viral RNA was quantified in the brain and cerebrospinal fluid by RT-qPCR assays targeting SIV Gag. Neuroinflammation was assessed by flow cytometry and multiplex ELISA assays. Results RNA sequencing and flow cytometry data demonstrated immune surveillance of the rhesus CNS by innate and adaptive immune cells during homeostasis. Following SHIV infection, viral entry and integration within multiple brain regions demonstrated vulnerabilities of key cognitive and motor function brain regions to HIV-1 during the acute phase of infection. SHIV-induced transcriptional alterations were concentrated to the PFCw and STS with upregulation of gene expression pathways controlling innate and T-cell inflammatory responses. Within the PFCw, gene modules regulating microglial activation and T cell differentiation were induced at 28 days post-SHIV infection, with evidence for stimulation of immune effector programs characteristic of neuroinflammation. Furthermore, enrichment of pathways regulating mitochondrial respiratory capacity, synapse assembly, and oxidative and endoplasmic reticulum stress were observed. These acute neuroinflammatory features were substantiated by increased influx of activated T cells into the CNS. Conclusions Our data show pervasive immune surveillance of the rhesus CNS at homeostasis and reveal perturbations of important immune, neuronal, and synaptic pathways within key anatomic regions controlling cognition and motor function during acute HIV infection. These findings provide a valuable framework to understand early molecular features of HIV associated neurodegeneration. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02610-y.
Collapse
Affiliation(s)
- Chase E Hawes
- Graduate Group in Immunology, University of California, Davis, CA, 95616, USA.,Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA
| | - Sonny R Elizaldi
- Graduate Group in Immunology, University of California, Davis, CA, 95616, USA.,Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA
| | - Danielle Beckman
- California National Primate Research Center, University of California, Davis, CA, 95616, USA
| | - Giovanne B Diniz
- California National Primate Research Center, University of California, Davis, CA, 95616, USA
| | | | - Sean Ott
- California National Primate Research Center, University of California, Davis, CA, 95616, USA
| | - Blythe P Durbin-Johnson
- Division of Biostatistics, School of Medicine, University of California, Davis, CA, 95616, USA
| | | | - Andrea Gompers
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA
| | - John H Morrison
- California National Primate Research Center, University of California, Davis, CA, 95616, USA. .,Department of Neurology, School of Medicine, University of California, Davis, CA, 95616, USA.
| | - Smita S Iyer
- Center for Immunology and Infectious Diseases, University of California, Davis, CA, 95616, USA. .,California National Primate Research Center, University of California, Davis, CA, 95616, USA. .,Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
14
|
Moysi E, Paris RM, Le Grand R, Koup RA, Petrovas C. Human lymph node immune dynamics as driver of vaccine efficacy: an understudied aspect of immune responses. Expert Rev Vaccines 2022; 21:633-644. [PMID: 35193447 DOI: 10.1080/14760584.2022.2045198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION During the last century, changes in hygiene, sanitation, and the advent of childhood vaccination have resulted in profound reductions in mortality from infectious diseases. Despite this success, infectious diseases remain an enigmatic public health threat, where effective vaccines for influenza, human immunodeficiency virus (HIV), tuberculosis, and malaria, among others remain elusive. AREA COVERED In addition to the immune evasion tactics employed by complex pathogens, our understanding of immunopathogenesis and the development of effective vaccines is also complexified by the inherent variability of human immune responses. Lymph nodes (LNs) are the anatomical sites where B cell responses develop. An important, but understudied component of immune response complexity is variation in LN immune dynamics and in particular variation in germinal center follicular helper T cells (Tfh) and B cells which can be impacted by genetic variation, aging, the microbiome and chronic infection. EXPERT OPINION This review describes the contribution of genetic variation, aging, microbiome and chronic infection on LN immune dynamics and associated Tfh responses and offers perspective on how inclusion of LN immune subset and cytoarchitecture analyses, along with peripheral blood biomarkers can supplement systems vaccinology or immunology approaches for the development of vaccines or other interventions to prevent infectious diseases.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | | | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA.,Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Roth GA, Picece VCTM, Ou BS, Luo W, Pulendran B, Appel EA. Designing spatial and temporal control of vaccine responses. NATURE REVIEWS. MATERIALS 2022; 7:174-195. [PMID: 34603749 PMCID: PMC8477997 DOI: 10.1038/s41578-021-00372-2] [Citation(s) in RCA: 142] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 05/02/2023]
Abstract
Vaccines are the key technology to combat existing and emerging infectious diseases. However, increasing the potency, quality and durability of the vaccine response remains a challenge. As our knowledge of the immune system deepens, it becomes clear that vaccine components must be in the right place at the right time to orchestrate a potent and durable response. Material platforms, such as nanoparticles, hydrogels and microneedles, can be engineered to spatially and temporally control the interactions of vaccine components with immune cells. Materials-based vaccination strategies can augment the immune response by improving innate immune cell activation, creating local inflammatory niches, targeting lymph node delivery and controlling the time frame of vaccine delivery, with the goal of inducing enhanced memory immunity to protect against future infections. In this Review, we highlight the biological mechanisms underlying strong humoral and cell-mediated immune responses and explore materials design strategies to manipulate and control these mechanisms.
Collapse
Affiliation(s)
- Gillie A. Roth
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Vittoria C. T. M. Picece
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- Department of Chemistry and Applied Biosciences, ETH Zürich, Zürich, Switzerland
| | - Ben S. Ou
- Department of Bioengineering, Stanford University, Stanford, CA USA
| | - Wei Luo
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA USA
- Program in Immunology, Stanford University School of Medicine, Stanford, CA USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA USA
| | - Eric A. Appel
- Department of Bioengineering, Stanford University, Stanford, CA USA
- Department of Materials Science & Engineering, Stanford University, Stanford, CA USA
- ChEM-H Institute, Stanford University, Stanford, CA USA
- Department of Paediatrics — Endocrinology, Stanford University School of Medicine, Stanford, CA USA
| |
Collapse
|
16
|
Oyong DA, Loughland JR, Soon MSF, Chan JA, Andrew D, Wines BD, Hogarth PM, Olver SD, Collinge AD, Varelias A, Beeson JG, Kenangalem E, Price RN, Anstey NM, Minigo G, Boyle MJ. Adults with Plasmodium falciparum malaria have higher magnitude and quality of circulating T-follicular helper cells compared to children. EBioMedicine 2022; 75:103784. [PMID: 34968760 PMCID: PMC8718734 DOI: 10.1016/j.ebiom.2021.103784] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 11/28/2021] [Accepted: 12/11/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Protective malarial antibodies are acquired more rapidly in adults than children, independently of cumulative exposure, however the cellular responses mediating these differences are unknown. CD4 T-follicular helper (Tfh) cells have key roles in inducing antibodies, with Th2-Tfh cell activation associated with antibody development in malaria. Whether Tfh cell activation in malaria is age dependent is unknown and no studies have compared Tfh cell activation in children and adults with malaria. METHODS We undertook a comprehensive study of Tfh cells, along with B cells and antibody induction in children and adults with malaria. Activation and proliferation of circulating Tfh (cTfh) cell subsets was measured ex vivo and parasite-specific Tfh cell frequencies and functions studied with Activation Induced Marker (AIM) assays and intracellular cytokine staining. FINDINGS During acute malaria, the magnitude of cTfh cell activation was higher in adults than in children and occurred across all cTfh cell subsets in adults but was restricted only to the Th1-cTfh subset in children. Further, adults had higher levels of parasite-specific cTfh cells, and cTfh cells which produced more Th2-Tfh associated cytokine IL-4. Consistent with a role of higher Tfh cell activation in rapid immune development in adults, adults had higher activation of B cells during infection and higher induction of antibodies 7 and 28 days after malaria compared to children. INTERPRETATION Our data provide evidence that age impacts Tfh cell activation during malaria, and that these differences may influence antibody induction after treatment. Findings have important implications for vaccine development in children. FUNDING This word was supported by the National Health and Medical Research Council of Australia, Wellcome Trust, Charles Darwin University Menzies School of Health Research, Channel 7 Children's Research Foundation, and National Health Institute.
Collapse
Affiliation(s)
- Damian A Oyong
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia; Charles Darwin University, Darwin, NT, Australia
| | - Jessica R Loughland
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Megan S F Soon
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jo-Anne Chan
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology, Central Clinical School, Monash University, VIC, Australia; Department of Medicine, University of Melbourne, VIC, Australia
| | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Bruce D Wines
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology, Central Clinical School, Monash University, VIC, Australia; Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - P Mark Hogarth
- Burnet Institute, Melbourne, VIC, Australia; Department of Immunology, Central Clinical School, Monash University, VIC, Australia; Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Stuart D Olver
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Alika D Collinge
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Antiopi Varelias
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, QLD, Australia
| | - James G Beeson
- Burnet Institute, Melbourne, VIC, Australia; Department of Medicine, University of Melbourne, VIC, Australia; Department of Microbiology, Monash University, VIC, Australia
| | - Enny Kenangalem
- Timika Malaria Research Program, Papuan Health and Community Development Foundation, Timika, Papua, Indonesia; District Health Authority, Timika, Papua, Indonesia
| | - Ric N Price
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia; Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - Gabriela Minigo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia; Charles Darwin University, Darwin, NT, Australia
| | - Michelle J Boyle
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia; QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Burnet Institute, Melbourne, VIC, Australia; Faculty of Medicine, The University of Queensland, QLD, Australia.
| |
Collapse
|
17
|
Deere JD, Carroll TD, Dutra J, Fritts L, Sammak RL, Yee JL, Olstad KJ, Reader JR, Kistler A, Kamm J, Di Germanio C, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Simmons G, Watanabe J, Pollard RE, Usachenko J, Immareddy R, Schmidt BA, O’Connor SL, DeRisi J, Busch MP, Iyer SS, Van Rompay KKA, Hartigan-O’Connor DJ, Miller CJ. SARS-CoV-2 Infection of Rhesus Macaques Treated Early with Human COVID-19 Convalescent Plasma. Microbiol Spectr 2021; 9:e0139721. [PMID: 34817208 PMCID: PMC8612156 DOI: 10.1128/spectrum.01397-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Human clinical studies investigating use of convalescent plasma (CP) for treatment of coronavirus disease 2019 (COVID-19) have produced conflicting results. Outcomes in these studies may vary at least partly due to different timing of CP administration relative to symptom onset. The mechanisms of action of CP include neutralizing antibodies but may extend beyond virus neutralization to include normalization of blood clotting and dampening of inflammation. Unresolved questions include the minimum therapeutic titer in the CP units or CP recipient as well as the optimal timing of administration. Here, we show that treatment of macaques with CP within 24 h of infection does not reduce viral shedding in nasal or lung secretions compared to controls and does not detectably improve any clinical endpoint. We also demonstrate that CP administration does not impact viral sequence diversity in vivo, although the selection of a viral sequence variant in both macaques receiving normal human plasma was suggestive of immune pressure. Our results suggest that CP, administered to medium titers, has limited efficacy, even when given very early after infection. Our findings also contribute information important for the continued development of the nonhuman primate model of COVID-19. These results should inform interpretation of clinical studies of CP in addition to providing insights useful for developing other passive immunotherapies and vaccine strategies. IMPORTANCE Antiviral treatment options for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain very limited. One treatment that was explored beginning early in the pandemic (and that is likely to be tested early in future pandemics) is plasma collected from people who have recovered from coronavirus disease 2019 (COVID-19), known as convalescent plasma (CP). We tested if CP reduces viral shedding or disease in a nonhuman primate model. Our results demonstrate that administration of CP 1 day after SARS-CoV-2 infection had no significant impact on viral loads, clinical disease, or sequence diversity, although treatment with normal human plasma resulted in selection of a specific viral variant. Our results demonstrate that passive immunization with CP, even during early infection, provided no significant benefit in a nonhuman primate model of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jesse D. Deere
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Timothy D. Carroll
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Joseph Dutra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Linda Fritts
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Rebecca Lee Sammak
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - JoAnn L. Yee
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Katherine J. Olstad
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - J. Rachel Reader
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Amy Kistler
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Jack Kamm
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | | | | - Sonny R. Elizaldi
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Jamin W. Roh
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Rachel E. Pollard
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Jodie Usachenko
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Brian A. Schmidt
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph DeRisi
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Smita S. Iyer
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Dennis J. Hartigan-O’Connor
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Christopher J. Miller
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
18
|
Scaria PV, Anderson C, Muratova O, Alani N, Trinh HV, Nadakal ST, Zaidi I, Lambert L, Beck Z, Barnafo EK, Rausch KM, Rowe C, Chen B, Matyas GR, Rao M, Alving CR, Narum DL, Duffy PE. Malaria transmission-blocking conjugate vaccine in ALFQ adjuvant induces durable functional immune responses in rhesus macaques. NPJ Vaccines 2021; 6:148. [PMID: 34887448 PMCID: PMC8660773 DOI: 10.1038/s41541-021-00407-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
Malaria transmission-blocking vaccines candidates based on Pfs25 and Pfs230 have advanced to clinical studies. Exoprotein A (EPA) conjugate of Pfs25 in Alhydrogel® developed functional immunity in humans, with limited durability. Pfs230 conjugated to EPA (Pfs230D1-EPA) with liposomal adjuvant AS01 is currently in clinical trials in Mali. Studies with these conjugates revealed that non-human primates are better than mice to recapitulate the human immunogenicity and functional activity. Here, we evaluated the effect of ALFQ, a liposomal adjuvant consisting of TLR4 agonist and QS21, on the immunogenicity of Pfs25-EPA and Pfs230D1-EPA in Rhesus macaques. Both conjugates generated strong antibody responses and functional activity after two vaccinations though activity declined rapidly. A third vaccination of Pfs230D1-EPA induced functional activity lasting at least 9 months. Antibody avidity increased with each vaccination and correlated strongly with functional activity. IgG subclass analysis showed induction of Th1 and Th2 subclass antibody levels that correlated with activity.
Collapse
Affiliation(s)
- Puthupparampil V. Scaria
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Charles Anderson
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Olga Muratova
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Nada Alani
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Hung V. Trinh
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA
| | - Steven T. Nadakal
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Irfan Zaidi
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Lynn Lambert
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Zoltan Beck
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA ,grid.201075.10000 0004 0614 9826Henry M. Jackson Foundation for the Advancement of Military Medicine, 6720A Rockledge Drive, Bethesda, MD 20817 USA ,grid.410513.20000 0000 8800 7493Present Address: Pfizer, Vaccine Research and Development, Pearl River, NY USA
| | - Emma K. Barnafo
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Kelly M. Rausch
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Chris Rowe
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Beth Chen
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Gary R. Matyas
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - Mangala Rao
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - Carl R. Alving
- grid.507680.c0000 0001 2230 3166U.S. Military HIV Research Program, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910 USA
| | - David L. Narum
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| | - Patrick E. Duffy
- grid.419681.30000 0001 2164 9667Laboratory of Malaria Immunology and Vaccinology, NIAID/NIH, 29 Lincoln Drive, Building 29B, Bethesda, MD 20892-2903 USA
| |
Collapse
|
19
|
Zoldan K, Ehrlich S, Killmer S, Wild K, Smits M, Russ M, Globig AM, Hofmann M, Thimme R, Boettler T. Th1-Biased Hepatitis C Virus-Specific Follicular T Helper-Like Cells Effectively Support B Cells After Antiviral Therapy. Front Immunol 2021; 12:742061. [PMID: 34659236 PMCID: PMC8514946 DOI: 10.3389/fimmu.2021.742061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/13/2021] [Indexed: 11/13/2022] Open
Abstract
Circulating Th1-biased follicular T helper (cTfh1) cells have been associated with antibody responses to viral infection and after vaccination but their B cell helper functionality is less understood. After viral elimination, Tfh1 cells are the dominant subset within circulating Hepatitis C Virus (HCV)-specific CD4 T cells, but their functional capacity is currently unknown. To address this important point, we established a clone-based system to evaluate CD4 T cell functionality in vitro to overcome experimental limitations associated with their low frequencies. Specifically, we analyzed the transcription factor expression, cytokine secretion and B cell help in co-culture assays of HCV- (n = 18) and influenza-specific CD4 T cell clones (n = 5) in comparison to Tfh (n = 26) and Th1 clones (n = 15) with unknown antigen-specificity derived from healthy donors (n = 4) or direct-acting antiviral (DAA)-treated patients (n = 5). The transcription factor expression and cytokine secretion patterns of HCV-specific CD4 T cell clones indicated a Tfh1 phenotype, with expression of T-bet and Bcl6 and production of IFN-γ and IL-21. Their B helper capacity was superior compared to influenza-specific or Tfh and Th1 clones. Moreover, since Tfh cells are enriched in the IFN-rich milieu of the HCV-infected liver, we investigated the impact of IFN exposure on Tfh phenotype and function. Type I IFN exposure was able to introduce similar phenotypic and functional characteristics in the Tfh cell population within PBMCs or Tfh clones in vitro in line with our finding that Tfh cells are elevated in HCV-infected patients shortly after initiation of IFN-α therapy. Collectively, we were able to functionally characterize HCV-specific CD4 T cells in vitro and not only confirmed a Tfh1 phenotype but observed superior Tfh functionality despite their Th1 bias. Furthermore, our results suggest that chronic type I IFN exposure supports the enrichment of highly functional HCV-specific Tfh-like cells during HCV infection. Thus, HCV-specific Tfh-like cells after DAA therapy may be a promising target for future vaccination design aiming to introduce a neutralizing antibody response.
Collapse
Affiliation(s)
- Katharina Zoldan
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Ehrlich
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Saskia Killmer
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katharina Wild
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Chemistry and Pharmacy, University of Freiburg, Freiburg, Germany
| | - Maike Smits
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Marissa Russ
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Chemistry and Pharmacy, University of Freiburg, Freiburg, Germany
| | - Anna-Maria Globig
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, University Hospital Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
20
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
21
|
Lakshmanappa YS, Roh JW, Rane NN, Dinasarapu AR, Tran DD, Velu V, Sheth AN, Ofotokun I, Amara RR, Kelley CF, Waetjen E, Iyer SS. Circulating integrin α 4 β 7 + CD4 T cells are enriched for proliferative transcriptional programs in HIV infection. FEBS Lett 2021; 595:2257-2270. [PMID: 34278574 DOI: 10.1002/1873-3468.14163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/13/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022]
Abstract
HIV preferentially infects α4 β7 + CD4 T cells, forming latent reservoirs that contribute to HIV persistence during antiretroviral therapy. However, the properties of α4 β7 + CD4 T cells in blood and mucosal compartments remain understudied. Employing two distinct models of HIV infection, HIV-infected humans and simian-human immunodeficiency virus (SHIV)-infected rhesus macaques, we show that α4 β7 + CD4 T cells in blood are enriched for genes regulating cell cycle progression and cellular metabolism. Unlike their circulating counterparts, rectal α4 β7 + CD4 T cells exhibited a core tissue-residency gene expression program. These features were conserved across primate species, indicating that the environment influences memory T-cell transcriptional networks. Our findings provide an important molecular foundation for understanding the role of α4 β7 in HIV infection.
Collapse
Affiliation(s)
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, UC Davis, CA, USA.,Graduate Group in Immunology, UC Davis, CA, USA
| | - Niharika N Rane
- Center for Immunology and Infectious Diseases, UC Davis, CA, USA
| | | | - Daphne D Tran
- Center for Immunology and Infectious Diseases, UC Davis, CA, USA
| | - Vijayakumar Velu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA.,Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA
| | - Anandi N Sheth
- Grady Infectious Diseases Program, Grady Health System, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Igho Ofotokun
- Grady Infectious Diseases Program, Grady Health System, Atlanta, GA, USA.,Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Rama R Amara
- Division of Microbiology and Immunology, Yerkes National Primate Research Center, Emory Vaccine Center, Emory University, Atlanta, GA, USA.,Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA, USA
| | - Colleen F Kelley
- Division of Infectious Diseases, Department of Medicine, The Hope Clinic of the Emory Vaccine Research Center, Emory University School of Medicine, Decatur, GA, USA
| | - Elaine Waetjen
- Department of Obstetrics and Gynecology, UC Davis School of Medicine, CA, USA
| | - Smita S Iyer
- Center for Immunology and Infectious Diseases, UC Davis, CA, USA.,California National Primate Research Center, UC Davis, CA, USA.,Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA, USA
| |
Collapse
|
22
|
Pino M, Abid T, Pereira Ribeiro S, Edara VV, Floyd K, Smith JC, Latif MB, Pacheco-Sanchez G, Dutta D, Wang S, Gumber S, Kirejczyk S, Cohen J, Stammen RL, Jean SM, Wood JS, Connor-Stroud F, Pollet J, Chen WH, Wei J, Zhan B, Lee J, Liu Z, Strych U, Shenvi N, Easley K, Weiskopf D, Sette A, Pollara J, Mielke D, Gao H, Eisel N, LaBranche CC, Shen X, Ferrari G, Tomaras GD, Montefiori DC, Sekaly RP, Vanderford TH, Tomai MA, Fox CB, Suthar MS, Kozlowski PA, Hotez PJ, Paiardini M, Bottazzi ME, Kasturi SP. A yeast expressed RBD-based SARS-CoV-2 vaccine formulated with 3M-052-alum adjuvant promotes protective efficacy in non-human primates. Sci Immunol 2021; 6:eabh3634. [PMID: 34266981 PMCID: PMC9119307 DOI: 10.1126/sciimmunol.abh3634] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/26/2021] [Accepted: 07/09/2021] [Indexed: 12/15/2022]
Abstract
Ongoing SARS-CoV-2 vaccine development is focused on identifying stable, cost-effective, and accessible candidates for global use, specifically in low and middle-income countries. Here, we report the efficacy of a rapidly scalable, novel yeast expressed SARS-CoV-2 specific receptor-binding domain (RBD) based vaccine in rhesus macaques. We formulated the RBD immunogen in alum, a licensed and an emerging alum adsorbed TLR-7/8 targeted, 3M-052-alum adjuvants. The RBD+3M-052-alum adjuvanted vaccine promoted better RBD binding and effector antibodies, higher CoV-2 neutralizing antibodies, improved Th1 biased CD4+T cell reactions, and increased CD8+ T cell responses when compared to the alum-alone adjuvanted vaccine. RBD+3M-052-alum induced a significant reduction of SARS-CoV-2 virus in respiratory tract upon challenge, accompanied by reduced lung inflammation when compared with unvaccinated controls. Anti-RBD antibody responses in vaccinated animals inversely correlated with viral load in nasal secretions and BAL. RBD+3M-052-alum blocked a post SARS-CoV-2 challenge increase in CD14+CD16++ intermediate blood monocytes, and Fractalkine, MCP-1, and TRAIL in the plasma. Decreased plasma analytes and intermediate monocyte frequencies correlated with reduced nasal and BAL viral loads. Lastly, RBD-specific plasma cells accumulated in the draining lymph nodes and not in the bone marrow, contrary to previous findings. Together, these data show that a yeast expressed, RBD-based vaccine+3M-052-alum provides robust immune responses and protection against SARS-CoV-2, making it a strong and scalable vaccine candidate.
Collapse
Affiliation(s)
- Maria Pino
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Talha Abid
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Susan Pereira Ribeiro
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Venkata Viswanadh Edara
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Katharine Floyd
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, U.S.A
| | - Muhammad Bilal Latif
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Gabriela Pacheco-Sanchez
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Debashis Dutta
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Shelly Wang
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Sanjeev Gumber
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Shannon Kirejczyk
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
- Division of Pathology, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce Cohen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rachelle L Stammen
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Sherrie M Jean
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer S Wood
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Fawn Connor-Stroud
- Division of Animal Resources, Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jeroen Pollet
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Wen-Hsiang Chen
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Junfei Wei
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Bin Zhan
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Jungsoon Lee
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Zhuyun Liu
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Ulrich Strych
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Neeta Shenvi
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, U.S.A
| | - Kirk Easley
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, U.S.A
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Justin Pollara
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Dieter Mielke
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Hongmei Gao
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Nathan Eisel
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Celia C LaBranche
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Xiaoying Shen
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - David C Montefiori
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center Durham, NC, USA
| | - Rafick P Sekaly
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Thomas H Vanderford
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
| | - Mark A Tomai
- 3M Corporate Research Materials Laboratory, St. Paul, MN, USA
| | | | - Mehul S Suthar
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A
- Emory Vaccine Center at Emory University, 954, Gatewood Rd, Atlanta, GA, U.S.A
- Centers for Childhood Infections and Vaccines; Children's Healthcare of Atlanta and Emory University, Department of Pediatrics, Atlanta, GA, U.S.A
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, U.S.A
| | - Peter J Hotez
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| | - Maria Elena Bottazzi
- Texas Children's Center for Vaccine Development, Houston, TX, U.S.A.
- Department of Pediatrics, Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, U.S.A
| | - Sudhir Pai Kasturi
- Division of Microbiology and Immunology, Yerkes National Primate Research Center at Emory University, 954 Gatewood Rd, Atlanta, GA, U.S.A.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, U.S.A
| |
Collapse
|
23
|
Soon MSF, Nalubega M, Boyle MJ. T-follicular helper cells in malaria infection and roles in antibody induction. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab008. [PMID: 36845571 PMCID: PMC9914587 DOI: 10.1093/oxfimm/iqab008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 01/29/2023] Open
Abstract
Immunity to malaria is mediated by antibodies that block parasite replication to limit parasite burden and prevent disease. Cytophilic antibodies have been consistently shown to be associated with protection, and recent work has improved our understanding of the direct and Fc-mediated mechanisms of protective antibodies. Antibodies also have important roles in vaccine-mediated immunity. Antibody induction is driven by the specialized CD4+ T cells, T-follicular helper (Tfh) cells, which function within the germinal centre to drive B-cell activation and antibody induction. In humans, circulating Tfh cells can be identified in peripheral blood and are differentiated into subsets that appear to have pathogen/vaccination-specific roles in antibody induction. Tfh cell responses are essential for protective immunity from Plasmodium infection in murine models of malaria. Our understanding of the activation of Tfh cells during human malaria infection and the importance of different Tfh cell subsets in antibody development is still emerging. This review will discuss our current knowledge of Tfh cell activation and development in malaria, and the potential avenues and pitfalls of targeting Tfh cells to improve malaria vaccines.
Collapse
Affiliation(s)
- Megan S F Soon
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia
| | - Mayimuna Nalubega
- Infectious Diseases Research Collaboration, Tororo District Hospital, Tororo, Uganda
| | - Michelle J Boyle
- Department of Infectious Diseases, QIMR-Berghofer, 300 Herston Road, Herston, QLD, 4006, Australia,Correspondence address. QIMR Berghofer Medical Research Institute, Brisbane, Australia. E-mail:
| |
Collapse
|
24
|
Ratnapriya S, Perez-Greene E, Schifanella L, Herschhorn A. Adjuvant-mediated enhancement of the immune response to HIV vaccines. FEBS J 2021; 289:3317-3334. [PMID: 33705608 DOI: 10.1111/febs.15814] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/28/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Protection from human immunodeficiency virus (HIV) acquisition will likely require an effective vaccine that elicits antibodies against the HIV-1 envelope glycoproteins (Envs), which are the sole target of neutralizing antibodies and a main focus of vaccine development. Adjuvants have been widely used to augment the magnitude and longevity of the adaptive immune responses to immunizations with HIV-1 Envs and to guide the development of specific immune responses. Here, we review the adjuvants that have been used in combination with HIV-1 Envs in several preclinical and human clinical trials in recent years. We summarize the interactions between the HIV-1 Envs and adjuvants, and highlight the routes of vaccine administration for various formulations. We then discuss the use of combinations of different adjuvants, the potential effect of adjuvants on the elicitation of antibodies enriched in somatic hypermutation and containing long complementarity-determining region 3 of the antibody heavy chain, and the elicitation of non-neutralizing antibodies.
Collapse
Affiliation(s)
- Sneha Ratnapriya
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Eva Perez-Greene
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Luca Schifanella
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Alon Herschhorn
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.,Microbiology, Immunology, and Cancer Biology Graduate Program, University of Minnesota, Minneapolis, MN, USA.,The College of Veterinary Medicine Graduate Program, University of Minnesota, Minneapolis, MN, USA.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
25
|
Helmold Hait S, Hogge CJ, Rahman MA, Hunegnaw R, Mushtaq Z, Hoang T, Robert-Guroff M. T FH Cells Induced by Vaccination and Following SIV Challenge Support Env-Specific Humoral Immunity in the Rectal-Genital Tract and Circulation of Female Rhesus Macaques. Front Immunol 2021; 11:608003. [PMID: 33584682 PMCID: PMC7876074 DOI: 10.3389/fimmu.2020.608003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
T follicular helper (TFH) cells are pivotal in lymph node (LN) germinal center (GC) B cell affinity maturation. Circulating CXCR5+ CD4+ T (cTFH) cells have supported memory B cell activation and broadly neutralizing antibodies in HIV controllers. We investigated the contribution of LN SIV-specific TFH and cTFH cells to Env-specific humoral immunity in female rhesus macaques following a mucosal Ad5hr-SIV recombinant priming and SIV gp120 intramuscular boosting vaccine regimen and following SIV vaginal challenge. TFH and B cells were characterized by flow cytometry. B cell help was evaluated in TFH-B cell co-cultures and by real-time PCR. Vaccination induced Env-specific TFH and Env-specific memory (ESM) B cells in LNs. LN Env-specific TFH cells post-priming and GC ESM B cells post-boosting correlated with rectal Env-specific IgA titers, and GC B cells at the same timepoints correlated with vaginal Env-specific IgG titers. Vaccination expanded cTFH cell responses, including CD25+ Env-specific cTFH cells that correlated negatively with vaginal Env-specific IgG titers but positively with rectal Env-specific IgA titers. Although cTFH cells post-2nd boost positively correlated with viral-loads following SIV challenge, cTFH cells of SIV-infected and protected macaques supported maturation of circulating B cells into plasma cells and IgA release in co-culture. Additionally, cTFH cells of naïve macaques promoted upregulation of genes associated with B cell proliferation, BCR engagement, plasma cell maturation, and antibody production, highlighting the role of cTFH cells in blood B cell maturation. Vaccine-induced LN TFH and GC B cells supported anti-viral mucosal immunity while cTFH cells provided B cell help in the periphery during immunization and after SIV challenge. Induction of TFH responses in blood and secondary lymphoid organs is likely desirable for protective efficacy of HIV vaccines.
Collapse
Affiliation(s)
- Sabrina Helmold Hait
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christopher James Hogge
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mohammad Arif Rahman
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ruth Hunegnaw
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zuena Mushtaq
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tanya Hoang
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
26
|
Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Schmidt BA, Carroll TD, Weaver KD, Smith JC, Verma A, Deere JD, Dutra J, Stone M, Franz S, Sammak RL, Olstad KJ, Rachel Reader J, Ma ZM, Nguyen NK, Watanabe J, Usachenko J, Immareddy R, Yee JL, Weiskopf D, Sette A, Hartigan-O'Connor D, McSorley SJ, Morrison JH, Tran NK, Simmons G, Busch MP, Kozlowski PA, Van Rompay KKA, Miller CJ, Iyer SS. SARS-CoV-2 induces robust germinal center CD4 T follicular helper cell responses in rhesus macaques. Nat Commun 2021; 12:541. [PMID: 33483492 PMCID: PMC7822826 DOI: 10.1038/s41467-020-20642-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/08/2020] [Indexed: 12/28/2022] Open
Abstract
CD4 T follicular helper (Tfh) cells are important for the generation of durable and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates Tfh cells and stimulates the germinal center (GC) response is an important question as we investigate vaccine induced immunity against COVID-19. Here, we report that SARS-CoV-2 infection in rhesus macaques, either infused with convalescent plasma, normal plasma, or receiving no infusion, resulted in transient accumulation of pro-inflammatory monocytes and proliferating Tfh cells with a Th1 profile in peripheral blood. CD4 helper cell responses skewed predominantly toward a Th1 response in blood, lung, and lymph nodes. SARS-CoV-2 Infection induced GC Tfh cells specific for the SARS-CoV-2 spike and nucleocapsid proteins, and a corresponding early appearance of antiviral serum IgG antibodies. Collectively, the data show induction of GC responses in a rhesus model of mild COVID-19.
Collapse
Affiliation(s)
| | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
- Graduate Group in Immunology, UC Davis, Davis, CA, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
- Graduate Group in Immunology, UC Davis, Davis, CA, USA
| | - Brian A Schmidt
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
| | - Timothy D Carroll
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Kourtney D Weaver
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Justin C Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Anil Verma
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
| | - Jesse D Deere
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Joseph Dutra
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Mars Stone
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Vitalant Research Institute, San Francisco, CA, USA
| | - Sergej Franz
- Vitalant Research Institute, San Francisco, CA, USA
| | | | | | - J Rachel Reader
- California National Primate Research Center, UC Davis, Davis, CA, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - Zhong-Min Ma
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Nancy K Nguyen
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
| | - Jennifer Watanabe
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Jodie Usachenko
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Ramya Immareddy
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - JoAnn L Yee
- California National Primate Research Center, UC Davis, Davis, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, San Diego, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, La Jolla, San Diego, CA, USA
| | - Dennis Hartigan-O'Connor
- California National Primate Research Center, UC Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, School of Medicine, UC Davis, Davis, CA, USA
| | - Stephen J McSorley
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - John H Morrison
- California National Primate Research Center, UC Davis, Davis, CA, USA
- Department of Neurology, School of Medicine, UC Davis, Davis, CA, USA
| | - Nam K Tran
- Pathology and Laboratory Medicine, School of Medicine, UC Davis, Davis, CA, USA
| | - Graham Simmons
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Vitalant Research Institute, San Francisco, CA, USA
| | - Michael P Busch
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
- Vitalant Research Institute, San Francisco, CA, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, UC Davis, Davis, CA, USA.
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, CA, USA.
| | - Christopher J Miller
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA.
- California National Primate Research Center, UC Davis, Davis, CA, USA.
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, CA, USA.
| | - Smita S Iyer
- Center for Immunology and Infectious Diseases, UC Davis, Davis, CA, USA.
- California National Primate Research Center, UC Davis, Davis, CA, USA.
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, Davis, CA, USA.
| |
Collapse
|
27
|
Th2-like T Follicular Helper Cells Promote Functional Antibody Production during Plasmodium falciparum Infection. CELL REPORTS MEDICINE 2020; 1:100157. [PMID: 33377128 PMCID: PMC7762767 DOI: 10.1016/j.xcrm.2020.100157] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/08/2020] [Accepted: 11/19/2020] [Indexed: 01/10/2023]
Abstract
CD4+ T follicular helper cells (Tfh) are key drivers of antibody development. During Plasmodium falciparum malaria in children, the activation of Tfh is restricted to the Th1 subset and not associated with antibody levels. To identify Tfh subsets that are associated with antibody development in malaria, we assess Tfh and antibodies longitudinally in human volunteers with experimental P. falciparum infection. Tfh cells activate during infection, with distinct dynamics in different Tfh subsets. Th2-Tfh cells activate early, during peak infection, while Th1-Tfh cells activate 1 week after peak infection and treatment. Th2-Tfh cell activation is associated with the functional breadth and magnitude of parasite antibodies. In contrast, Th1-Tfh activation is not associated with antibody development but instead with plasma cells, which have previously been shown to play a detrimental role in the development of long-lived immunity. Thus, our study identifies the contrasting roles of Th2 and Th1-Tfh cells during experimental P. falciparum malaria.
Collapse
|
28
|
CD4 + T Cells Induced by Tuberculosis Subunit Vaccine H1 Can Improve the HIV-1 Env Humoral Response by Intrastructural Help. Vaccines (Basel) 2020; 8:vaccines8040604. [PMID: 33066267 PMCID: PMC7711721 DOI: 10.3390/vaccines8040604] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 01/25/2023] Open
Abstract
The induction of a potent and long-lasting, broadly neutralizing antibody response is one of the most promising approaches in HIV-1 vaccination. Recently, we demonstrated that Gag-specific T helper cells induced by DNA priming can enhance and modulate the HIV Env-specific B cell response upon virus-like particle (VLP) boost by intrastructural help (ISH). In order to minimize the induction of potentially harmful HIV specific TH cells, we explored the possibility to harness the heterologous TH cells induced by a recombinant tuberculosis subunit vaccine H1, which contains a fusion protein of Ag85B and ESAT-6 antigens in combination with the liposomal adjuvant CAF01. To provide ISH, immunodominant MHC-II restricted peptides from the H1 vaccine were genetically incorporated into the HIV 1 Gag protein and used for HIV VLP production. ISH effects on Env-specific antibody levels and B cell differentiation were analyzed in mice primed against H1 and boosted with VLPs. In contrast to non-primed mice, a significant increase of Env-specific IgG levels for up to 26 weeks after the last immunization was observed. This increase was largely caused by elevated IgG2b and IgG2c levels in mice that received H1 priming. Additionally, ISH enhanced the frequency of Env-specific long-lived plasma cells in the bone marrow. In this study, we were able to demonstrate that a heterologous prime-boost regimen consisting of the H1 tuberculosis subunit vaccine and T helper epitope modified HIV-1 VLPs resulted in enhanced HIV Env antibody and B cell responses, mediated by intrastructural help.
Collapse
|
29
|
Elizaldi S, Lakshmanappa YS, Roh J, Schmidt B, Carroll T, Weaver K, Smith J, Deere J, Dutra J, Stone M, Franz S, Sammak R, Olstad K, Reader JR, Ma ZM, Nguyen N, Watanabe J, Usachenko J, Immareddy R, Yee J, Weiskopf D, Sette A, Hartigan-O'Connor D, McSorley S, Morrison J, Tran N, Simmons G, Busch M, Kozlowsk P, van Rompay K, Miller C, Iyer S. SARS-CoV-2 infection induces robust germinal center CD4 T follicular helper cell responses in rhesus macaques. RESEARCH SQUARE 2020:rs.3.rs-51545. [PMID: 32818217 PMCID: PMC7430596 DOI: 10.21203/rs.3.rs-51545/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
CD4 T follicular helper (T fh ) cells are important for the generation of durable and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates T fh cells and stimulates the germinal center response is an important question as we investigate vaccine options for the current pandemic. Here we report that SARS-CoV-2 infection resulted in transient accumulation of pro-inflammatory monocytes and proliferating T fh cells with a T h 1 profile in peripheral blood. CD4 helper cell responses were skewed predominantly toward a T h 1 response in blood, lung, and lymph nodes. We observed the generation of germinal center T fh cells specific for the SARS-CoV-2 spike (S) and nucleocapsid (N) proteins, and a corresponding early appearance of antiviral serum IgG antibodies. Our data suggest that a vaccine promoting T h 1-type T fh responses that target the S protein may lead to protective immunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Justin Smith
- Louisiana State University Health Sciences Center
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, CA
| | | | | | | | | | | | | | | | - Pamela Kozlowsk
- Louisiana State University Health Sciences Center New Orleans
| | | | | | - Smita Iyer
- University of California Davis School of Veterinary Medicine and California National Primate Research Center
| |
Collapse
|
30
|
Elizaldi SR, Lakshmanappa YS, Roh JW, Schmidt BA, Carroll TD, Weaver KD, Smith JC, Deere JD, Dutra J, Stone M, Sammak RL, Olstad KJ, Reader JR, Ma ZM, Nguyen NK, Watanabe J, Usachaenko J, Immareddy R, Yee JL, Weiskopf D, Sette A, Hartigan-O’Connor D, McSorley SJ, Morrison JH, Tran NK, Simmons G, Busch MP, Kozlowski PA, Van Rompay KK, Miller CJ, Iyer SS. SARS-CoV-2 infection induces germinal center responses with robust stimulation of CD4 T follicular helper cells in rhesus macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.07.191007. [PMID: 32676606 PMCID: PMC7359530 DOI: 10.1101/2020.07.07.191007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD4 T follicular helper (T fh ) cells are important for the generation of long-lasting and specific humoral protection against viral infections. The degree to which SARS-CoV-2 infection generates T fh cells and stimulates the germinal center response is an important question as we investigate vaccine options for the current pandemic. Here we report that, following infection with SARS-CoV-2, adult rhesus macaques exhibited transient accumulation of activated, proliferating T fh cells in their peripheral blood on a transitory basis. The CD4 helper cell responses were skewed predominantly toward a T h 1 response in blood, lung, and lymph nodes, reflective of the interferon-rich cytokine environment following infection. We also observed the generation of germinal center T fh cells specific for the SARS-CoV-2 spike (S) and nucleocapsid (N) proteins, and a corresponding early appearance of antiviral serum IgG antibodies but delayed or absent IgA antibodies. Our data suggest that a vaccine promoting Th1-type Tfh responses that target the S protein may lead to protective immunity.
Collapse
Affiliation(s)
- Sonny R. Elizaldi
- Center for Immunology and Infectious Diseases, UC Davis, CA
- Graduate Group in Immunology, UC Davis, CA
| | | | - Jamin W. Roh
- Center for Immunology and Infectious Diseases, UC Davis, CA
- Graduate Group in Immunology, UC Davis, CA
| | | | | | - Kourtney D. Weaver
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Justin C. Smith
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Jesse D. Deere
- California National Primate Research Center, UC Davis, CA
| | - Joseph Dutra
- California National Primate Research Center, UC Davis, CA
| | - Mars Stone
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA
- Vitalant Research Institute, San Francisco, CA
| | | | | | - J. Rachel Reader
- California National Primate Research Center, UC Davis, CA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA
| | - Zhong-Min Ma
- California National Primate Research Center, UC Davis, CA
| | | | | | | | | | - JoAnn L. Yee
- California National Primate Research Center, UC Davis, CA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA
| | - Dennis Hartigan-O’Connor
- California National Primate Research Center, UC Davis, CA
- Department of Medical Microbiology and Immunology, School of Medicine, UC Davis, CA
| | - Stephen J. McSorley
- Center for Immunology and Infectious Diseases, UC Davis, CA
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, UC Davis, CA
| | - John H. Morrison
- California National Primate Research Center, UC Davis, CA
- Department of Neurology, School of Medicine, UC Davis, CA
| | - Nam K. Tran
- Pathology and Laboratory Medicine, School of Medicine, UC Davis, CA
| | - Graham Simmons
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA
- Vitalant Research Institute, San Francisco, CA
| | - Michael P Busch
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA
- Vitalant Research Institute, San Francisco, CA
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA
| | - Koen K.A. Van Rompay
- California National Primate Research Center, UC Davis, CA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA
| | - Christopher J. Miller
- Center for Immunology and Infectious Diseases, UC Davis, CA
- California National Primate Research Center, UC Davis, CA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA
| | - Smita S. Iyer
- Center for Immunology and Infectious Diseases, UC Davis, CA
- California National Primate Research Center, UC Davis, CA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, UC Davis, CA
| |
Collapse
|